Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Cardioprotection of Controlled and Cardiac-Specific Over-Expression of A2A-Adenosine Receptor in the Pressure Overload

  • Eman A. Hamad,

    Affiliations Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America, Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America

  • Weizhong Zhu ,

    Arthur.Feldman@Temple.edu (AMF); Weizhong.Zhu@Temple.edu (WZ)

    Affiliation Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America

  • Tung O. Chan,

    Affiliation Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America

  • Valerie Myers,

    Affiliation Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America

  • Erhe Gao,

    Affiliation Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America

  • Xue Li,

    Affiliation Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America

  • Jin Zhang,

    Affiliation Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America

  • Jianliang Song,

    Affiliation Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America

  • Xue-Qian Zhang,

    Affiliation Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America

  • Joseph Y. Cheung,

    Affiliation Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America

  • Walter Koch,

    Affiliation Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America

  • Arthur M. Feldman

    Arthur.Feldman@Temple.edu (AMF); Weizhong.Zhu@Temple.edu (WZ)

    Affiliation Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America

Abstract

Adenosine binds to three G protein-coupled receptors (R) located on the cardiomyocyte (A1-R, A2A-R and A3-R) and provides cardiac protection during both ischemic and load-induced stress. While the role of adenosine receptor-subtypes has been well defined in the setting of ischemia-reperfusion, far less is known regarding their roles in protecting the heart during other forms of cardiac stress. Because of its ability to increase cardiac contractility and heart rate, we hypothesized that enhanced signaling through A2A-R would protect the heart during the stress of transverse aortic constriction (TAC). Using a cardiac-specific and inducible promoter, we selectively over-expressed A2A-R in FVB mice. Echocardiograms were obtained at baseline, 2, 4, 8, 12, 14 weeks and hearts were harvested at 14 weeks, when WT mice developed a significant decrease in cardiac function, an increase in end systolic and diastolic dimensions, a higher heart weight to body weight ratio (HW/BW), and marked fibrosis when compared with sham-operated WT. More importantly, these changes were significantly attenuated by over expression of the A2A-R. Furthermore, WT mice also demonstrated marked increases in the hypertrophic genes β-myosin heavy chain (β-MHC), and atrial natriuretic factor (ANF) – changes that are mediated by activation of the transcription factor GATA-4. Levels of the mRNAs encoding β-MHC, ANP, and GATA-4 were significantly lower in myocardium from A2A-R TG mice after TAC when compared with WT and sham-operated controls. In addition, three inflammatory factors genes encoding cysteine dioxygenase, complement component 3, and serine peptidase inhibitor, member 3N, were enhanced in WT TAC mice, but their expression was suppressed in A2A-R TG mice. A2A-R over-expression is protective against pressure-induced heart failure secondary to TAC. These cardioprotective effects are associated with attenuation of GATA-4 expression and inflammatory factors. The A2A-R may provide a novel new target for pharmacologic therapy in patients with cardiovascular disease.

Introduction

Adenosine is an endogenous purine nucleoside that plays an important role in protecting the heart during ischemia. The cardiovascular effects of adenosine (A) are mediated by 4 G-protein–coupled receptors (A1-R, A2A-R, A2B-R and A3-R), all of which are expressed in the heart. Activation of A2A -Rs results in coupling to Gs proteins and activation of adenylyl cyclase [1], [2], [3] while activation of the A1- and A3-Rs inhibits adenylyl cyclase and modulates other signaling pathways regulated by Gi/o. Studies using murine models in which the A1- and A3-Rs have been genetically manipulated demonstrate a critical role for these receptors in cardiac protection during ischemia and reperfusion. [4], [5] By contrast, A2A-Rs have been shown to promote post ischemic protection through inhibition of inflammatory responses. [6], [7].

Owing at least in part to its pharmacological effects on neurohormone and cytokine activation, [8], [9] adenosine also affects ventricular remodeling in models of heart failure. For example, adenosine attenuates detrimental chamber remodeling in rodents with pressure overload hypertrophy and decreases cell size in cultured neonatal cardiomyocytes. [10], [11], [12], [13] However, the role of adenosine receptor-subtypes in cardiac remodeling has not been fully elucidated. Pharmacologic activation of the A1-R effectively attenuated the development of cardiac hypertrophy and prevented heart failure in mice that underwent transverse aortic constriction (TAC) [11] and mice that were A1-R gene-deficient had a higher mortality when compared with wild-type controls but did not demonstrate altered ventricular hypertrophy or increased cardiac dysfunction. [14] Surprisingly, mice in which the A3-R had been knocked out demonstrated an improved survival, decreased fibrosis and hypertrophy and a more robust left ventricular function after TAC when compared with wild-type controls. The role of the A2-R in cardiac remodeling has not been defined.

Previously, we demonstrated that constitutive and cardiac specific over-expression of the A2A -R induced a hyper-contractile phenotype with enhanced calcium handling that prevented heart failure in a transgenic model [15]. This led us to hypothesize that signaling through the A2A -R might also have salutary effects on cardiac remodeling. To test this hypothesis we assessed the effects of TAC on cardiac morphology, function and gene expression in wild type mice and in mice with cardiac specific and controlled (adult) over-expression of the A2a-R.

Sustained myocardial hypertrophy secondary to pressure overload is a leading cause in the development of heart failure and sudden death in humans [16], [17]. Hemodynamic overload is a complex physiological stimulus that can lead to marked changes in myocardial structure and function through various humeral and mechanical components. The hypertrophic response induced by pressure overload is associated with marked alterations in cardiac gene expression, which include reactivation of fetal gene expression patterns. Many studies demonstrated an increase in the expression of the fetal gene beta myosin heavy chain (β-MHC) as a sensitive marker for hypertrophy [18]. Many signaling pathways have been implicated in cardiac hypertrophy and subsequent failure. GATA-4 a cardiac restricted zinc finger transcription factor has been shown to control several genes up regulated during cardiac hypertrophy including β-MHC, cardiac troponin-C, atrial natriuretic factor, sodium/calcium exchanger (NCX), A1-R [19]. With that said, not all hypertrophy is thought to be deleterious. Animal models of hypertrophy have demonstrated adaptive hypertrophy with normalized wall stress and full compensation. For example, Insulin like growth factor (IGF) which has a signaling system involving Protein kinase B (PKB) has been described in an adaptive pressure induced process [20]. Athletes are thought to have physiologic hypertrophy secondary to endurance training, which is not associated with fibrosis or up regulation in hypertrophic response genes, and increases in wall thickness are modest.

Results

We created mice with inducible overexpression of A2A-AR. The human A2A-AR cDNA was cloned into a cardiac-specific and inducible controlled vector (TREMHC) composed of a modified mouse α-myosin heavy chain (α-MHC) minimal promoter fused with nucleotide binding sites for tetracycline transactivating factor (tTA) (Fig. 1A). [21] A2A-AR transgenic (TG) mice were engineered on an FVB background (PolyGene, Zurich, Switzerland) and crossed with mice that expressed tTA in the heart (MHC-tTA; Fig. 1A). In this “tetracycline-off” inducible system, the stable tetracycline analog doxycycline (DOX) inhibits tTA transactivation, and it was administered to mice at 300 mg/kg of mouse diet (Bio-Serv, Frenchtown, NJ). A2A-R transgenic founder lines expressing low and high levels of A2A-R as shown in Figure 1B, as evidenced by western blot. The constitutive model was not placed on doxycycline, while the induced model was placed on doxycycline during mating and removed after 3 weeks (Fig. 1C). As seen in Figure 1D, A2A-R was really detectable at 6-week-old mice by 3 weeks of induction. Mice generation was confirmed in our previous studies [22], [23], [24].

thumbnail
Figure 1. Over-expression of the A2A adenosine receptor in mice myocardium.

Mice with constitutive and controlled overexpression of A2A-R were created. (A & B) Bi-transgenic, cardiac specific doxycyline regulated A2A-R transgenic mice were generated and confirmed there is A2A-R expression in all of lines; (C & D) A representative diagram of the timeline of gene induction. The constitutive model was not placed on doxycyline and over expressed A2A-R at birth while the controlled or induced model was placed on doxcycline during mating and removed at the age of 3 weeks.

https://doi.org/10.1371/journal.pone.0039919.g001

At eight weeks of age, A2A-R TG mice demonstrated a significant increase in fractional shortening by 15–20% compared with non-transgenic littermates (Fig. 2A, P<0.05, n = 12), but were otherwise phenotypically normal. In contrast, heart rates and wall thickness were significantly increased in constitutive expression of A2A-R mice [23]. The increase in fractional shortening persisted at 24 weeks of age. The systolic intracellular Ca2+ in cardiac myoyctes from the mice at 10–12 weeks of age was significantly enhanced as seen in Fig. 2B (p<0.05, 15 cells from 5 mice hearts). At the same time, the recovery of intracellular Ca2+ were markedly rapid as shown in Fig. 2C (p<0.05, 23 cells from 5 mice hearts).

thumbnail
Figure 2. Effects of cardiac specific A2A-R e

xpression on cardiac funtion and calcium handeling. (A) Echocardiography of mice with inducible, cardiac restricted expression of A2A-R TG and wild type (WT) mice. Fraction shorting (FS) at 8 week and 24 weeks in A2A-R TG and WT mice showed persist hyper-contractile phenotype in A2A-R TG mice up to 24 weeks (*p<0.01 vs WT mice, n = 8). (B & C) Calcium transient data showing increased systolic calcium (B) and rapid calcium re-uptake activity (C) in cardiomyocytes from the A2A-R TG mice at 10 weeks age compared to WT. Data were expressed as mean±SE. *p<0.01 compared to WT cardiomyocytes, n = 15 cells from 5 mice hearts.

https://doi.org/10.1371/journal.pone.0039919.g002

As expected, cardiac pressure overload by TAC caused a significant decrease in cardiac contractile function (Fig. 3A, Table 1) in WT mice. These changes could be seen as early as two weeks after TAC and persisted to the end experimental point at 14 weeks after TAC (p<0.001, n = 17, repeated measures two-way ANOVA test). The increase in end-systolic and end-diastolic dimension (Fig. 3B) and a higher heart weight to body weight ratio (HW/BW) (Fig. 3C) compared with sham-operated controls were attributed to the contractile dysfunction. More importantly, the development of left ventricular dysfunction (Fig. 3A & Table 2, p<0.01, n = 10–17), End systolic dimension (Fig. 3B & Table 2), heart/body ratio (Fig. 3C, p<0.01,n = 10–17), and cardiac fibrosis (Fig. 3D, p<0.01, n = 10–17) were markedly attenuated in mice with inducible, cardiac specific over-expressing A2A-R (Fig. 3A, 3B, 3C, 3D) mice at 14 weeks after TAC.

thumbnail
Figure 3. Effect of TAC on left ventricular hypertrophy and function as measured by Echocardiography.

(A) A2A-R mice had preserved cardiac function and showed tolerance to TAC-induced pressure overload. WT mice developed a significant decrease in cardiac contractile function at 14 weeks after TAC (*P<0.01, repeated measures ANOVA test, n = 17). Of note, contractile function in A2A-R TG mice was slightly decline after TAC. *p<0.01 vs WT TAC at the same time point. (B &C) WT mice developed into a significant increase in end systolic dimensions (B) and a higher heart weight to body weight ratio (C).*p<0.01 vs sham group, n = 8–17; #p<0.01 vs WT TAC, n = 10–17. (D) WT mice showed significantly more fibrosis than A2A-R TG at 14 weeks post TAC. Data was expressed as mean±SE *p<0.01 vs WT, n = 10–17. The basal fibrosis is no difference between WT and A2A-R TG. Both were below 0.1% of total myocardium area.

https://doi.org/10.1371/journal.pone.0039919.g003

thumbnail
Table 1. Effect of TAC on LV hypertrophy and Function as Measured by Echocardiography.

https://doi.org/10.1371/journal.pone.0039919.t001

To assess the effects of pressure overload on gene expression in A2A-R TG and WT mice with or without pressure overload, we measured mRNA levels of the hypertrophic response genes ß-MHC and ANF as well as the transcription factor GATA-4. As seen in Figure 4, it was indeed that hypertrophic marker genes, the mRNAs encoding ANP (Fig. 4A, p<0.05, n = 7) and ß-MHC (Fig. 4B, p<0.05, n = 7), were significantly enhanced by 40.5±5.8% and 70.7±3.5%, respectively, in WT mice TAC group compared to sham group. Of note, these hypertrophic marker genes were dramatically suppressed in the inducible, cardiac-specific A2A-R TG mice (Fig. 4A & 4B). In addition, the mRNA encoding GATA-4, a transcription factor that mediates the activation of the hypertrophic gene program was expressed at a significantly lower level in A2A-R TG mice than that in wild type littermate controls after TAC (Fig. 4C, p<0.01, n = 7–8). Since overexpression of A1-R is known to cause a decrease in cardiac function [15], we measured the A1-R mRNA levels in both WT and A2A-R TG mice. As expected, the WT mice had a significant increase in A1-R levels 14 weeks (p<0.001 vs sham, n = 6) after TAC, but not in A2A-R TG mice (p<0.01 vs WT TAC group, n = 6), as shown in Fig. 4D.

thumbnail
Figure 4. Effect of TAC on GATA-4, ANP, β-MHC and A1-R expression in A2A-R and WT mice.

After 14 weeks of TAC, the total RNA was isolated from either A2A-R TG or WT mice myocardium. The Q-PCR was performed to check the gene expression of GATA-4 ANP, β-MHC and A1-R. Data were expressed as mean ± SE. p<0.05 vs sham, #p<0.05 vs WT mice TAC group, n = 6.

https://doi.org/10.1371/journal.pone.0039919.g004

Since it has recently been shown that cardiac inflammation are one of the major pathological factors involving in the pressure overload-induced murine heart failure [25], [26], [27] and activation of A2A-R are responsible for its anti-inflammatory effects [28], [29], we screened the experimental mice myocardium by gene microarray and validated the gene changes found in microarray by Q-PCR. As shown in Figure 5, cysteine dioxygenase 1 (Cdo1), complement component 3 (C3), and serine (or cysteine) peptidase inhibitor, member 3N (Serpina3n) were enhanced in WT TAC mice, but their expression were suppressed in A2A-R TG mice. Interestingly, toll-like receptor (TLr 7), which synergize with A2A-R agonists and adenosine to up-regulate VEGF, while simultaneously strongly down-regulating TNFα expression [30], was increased in A2A-R TG mice even without TAC (Fig. 5D).

thumbnail
Figure 5. Effects of TAC on Inflammatory genes expression in A2A and WT Mice.

After 14wks of TAC, the total RNA was isolated from A2A-R TG and WT mice myocardium. The Q-PCR was performed to check the gene expression of cysteine dioxygenase 1 (Cdo1), complement component 3 (C3), serine (or cysteine) peptidase inhibitor, member 3N (Serpina3n), and toll-like receptor (TLr 7). Data was expressed as mean ± SE. *p<0.05 vs sham, #p<0.05 vs WT mice TAC group, n = 6. &p<0.05 vs WT sham.

https://doi.org/10.1371/journal.pone.0039919.g005

Discussion

The present study demonstrates for the first time that activation of the A2A-R signaling pathway can modulate the fibrosis, hypertrophy and subsequent left ventricular dysfunction that follow TAC using a murine model in which over-expression of the A2A receptor can be controlled and is cardiac specific. This model system provides several unique features. Enhanced expression of the A2A-R: (1) is cardiac specific, thereby obviating effects of adenosine receptor signaling in the peripheral vasculature or in the central nervous system; (2) can be “controlled” in order to preclude the known effects of adenosine receptor signaling on cardiac and neural development; and (3) avoids the potentially confounding effects of using non-selective or partially selective adenosine receptor pharmacologic agonists and antagonists.

In concentric hypertrophy induced by pressure overload, it has been suggested that myocytes grow in width to increase wall thickness in order to regulate the pressure induced by increased wall stress [31], [32]. With sustained volume load, the compensatory hypertrophy transitions to heart failure and dilation. Many mechanisms have been implicated in this transition including, increased collagen and fibrosis, an upset in the balance between metalloproteinases and their inhibitors, oxidative stress and neurohormal activation [33]. In the present study the WT mice developed more fibrosis than the A2A-R TG mice after TAC.

These salutary affects of enhanced A2A-R signaling were associated with a marked attenuation in the expression of the hypertrophy-associated genes β-MYC and ANF and the transcription regulatory protein GATA-4. β-MHC is characterized by low adenosine triphosphate activity and low filament sliding velocity but can generate cross-bridge force with higher economy of energy consumption [34], [35], [36]. This suggests that up regulation of β-MHC can be an early adaptive response to pressure overload but over time leads to a decrease in contractile function [37]. Indeed, Dorn et al, suggested that depressed myocyte contractility after induction of pressure overload hypertrophy in aortic banded FVB mice is due in part to transcriptional up regulation of β-MHC [38]. GATA-4 has been shown to control several genes up-regulated during cardiac hypertrophy including β-MHC and ANF [19]. GATA-4 binding sites are thought to be required for activation of β-MHC and angiotensin II type a receptor expression - both of which have been implicated in pathological ventricular hypertrophy [39] and the over expression of GATA-4 generated cardiac hypertrophy in cultured cardiomyocytes and in mice. [40], [41] Thus the finding that the diminished hypertrophy and failure after TAC in the A2A-R TG mice is associated with a decrease in GATA-4 expression may imply a link between A2A-R signaling and the expression of hypertrophy genes. GATA-4 expression level varies between tissues, between developmental stages, and in disease states. Although it is often used as a marker of cardiomyocytes, in fetal heart GATA-4 expression is the highest in the proepicardium followed by endocardial cushions and then cardiomyocytes. GATA-4 expression in the adult heart had been reported to increase by approximately twofold in heart disease [42], [43]. However, little is known about the regulatory sequences that drive cardiac GATA-4 expression. Interestingly, Gs-protein coupled β-AR promotes GATA-4 signaling associated with cardiac hypertrophy [44], [45], [46], [47]. By contrast, we reported here that A2A-R, another Gs protein-coupled receptors that also signals through activation of adenylate cyclase, appears to diminshe GATA-4 expression. However, by contrast with the β1-AR, the A2A-R can also mediate activation of MAPKs and PKC [48] with subsequent induction of hypoxia-inducible factor 1 [49]. Thus, it might be a PKA-independent pathway that suppresses GATA-4 expression in myocytes after A2A-R signaling. However, further studies will be required to test this hypothesis.

Earlier studies have suggested a role for adenosine in cardioprotection during pressure-induced stress. For example, treatment with dipyridamole, an adenosine uptake blocker that increases myocardial adenosine levels, attenuated chamber remodeling in rats with pressure overload hypertrophy. [10] Similarly, the adenosine analogue 2-chloroadenosine lowered both heart to body weight ratios and improved left ventricular fractional shortening in mice exposed to TAC. [10] Consistent with these studies, diminished extra-cellular adenosine production as a result of a genetic deletion of CD73 exacerbated left ventricular hypertrophy and dysfunction after pressure overload. [13] In vitro, all of three adenosine receptors blunt the phenylephrine-induced rat neonatal cardiomyocytes hypertrophy [12]. However, the role of the A1- and A3-adenosine receptors in protecting the heart from the stress of pressure overload remains less clear. Using the selective A1-adeonsine agonist N6-cyclopentyladeonsine (CPA), Liao et al found that A1-R signaling attenuated TAC-induced changes in left ventricular fractional shortening and heart to body-weight ratios in C57B6 mice. However, when the A1-R was genetically deleted, TAC had identical effects on ventricular hypertrophy and dysfunction. [14] Furthermore, deletion of the A3-R attenuated TAC-induced left ventricular hypertrophy, fibrosis and dysfunction, suggesting that over-expression of the A3-R would have a deleterious effect. Since A1- and A3-R signaling inhibit adenylyl cyclase, slow heart rate, and inhibit cardiac contractility while A2A-R signaling increases adenylyl cyclase activity and enhances cardiac contractility, it is not surprising that these different adenosine receptor-subtypes have disparate effects in the context of pressure-induced stress. [15].

A2A-R agonist displays rapid anti-inflammatory properties in a variety of in vitro and in vivo models [50], [51], [52]. And cardiac inflammation is one of the major pathological factors involving in the pressure overload-induced murine heart failure [25], [26], [27]. In the present study, four inflammatory factors are suppressed by over-expression of A2A-R, which might be attributable to its salutary effects on cardiac remodeling. Future studies will be required to determine why the enhanced cardiac specific A2A-R signaling suppresses myocardial inflammation and what the molecular relationship is between mycoytes, inflammatory cells, and fibroblast during enhanced A2A-R signaling.

In summary our study demonstrates that A2A-R over-expression is protective against pressure-induced heart failure secondary to TAC. These cardioprotective effects are associated with inhibition of GATA-4 expression and attenuation of the up-regulation of hypertrophy gene program that characterizes the pressure overloaded heart. Taken together, these results suggest that the A2A-R may be a therapeutic target in the treatment of patients with hypertension or hypertrophic heart disease.

Materials and Methods

Transgenic Mouse Generation

Experiments were carried out in transgenic mice with controlled cardiac restricted over expression of the human A2A-R TG as previously described [15]. Using a cardiac-specific and inducible promoter, we selectively over-expressed A2A-R TG in FVB mice after removal of doxycycline (DOX) from their diet at 3wks. Animal studies were approved by the Institutional Animal Care and Use Committee of Thomas Jefferson University.

[Ca2+]i Transient Measurements

Myocytes from A2A-R TG and WT mice were exposed to 0.67 µM of fura-2 AM for 15 minutes at 37°C. Fura-2-loaded myocytes were field-stimulated to contract (1 Hz, 37°C) in medium 199 containing 1.8 mM [Ca2+]o. Fura-2-loaded myocytes mounted on [Ca2+]i transient measurements using a Dvorak-Stotler chamber situated in a temperature-controlled stage (37°C) of a Zeiss IM 35 inverted microscope (Thornwood, NY) were performed as previously described [53].

Surgical Procedure for Transverse Aortic Banding

Eight-week-old male wild-type FVB mice (N = 17) and A2A -R littermates (N = 10) underwent transverse aortic banding (TAC) as previously described [15]. Briefly, an aortic band was created by placing a ligature (7-0 nylon suture) securely between the origin of the right innominate and left common carotid arteries with a 27-gauge needle as a guide. The sham procedure was identical except that the aorta was not ligated. Each strain has 8 mice in Sham group.

In Vivo Assessment of Cardiac Function

Left ventricular (LV) function was evaluated with transthoracic echocardiography at baseline, 2, 4, 8, 12, and 14Wks. A Visual Sonic Vevo 770 imaging system was used (Miami, FL). Mice were lightly sedated with isoflurane. A parasternal short-axis view was obtained for LV M-mode imaging at the papillary muscle level. Three independent M-mode images were used for measurements of LV end-diastolic internal diameter (LVEDD) and LV end-systolic internal diameter (LVESD) in two consecutive beats according to the American Society of Echocardiography leading edge method. Fractional shortening (FS) was calculated as FS%  = [(LVEDD – LVESD)/LVEDD]×100. Anterior (AWT) and Posterior Wall thickness (PWT) were also measured. Hearts were harvested at 14 weeks.

Real-Time Polymerase Chain Reaction

Reverse-transcribed cDNA from myocardial mRNA was used to determine the expression of A2-AR, atrial natriuretic peptide (ANP), GATA-4, and β-MHC. cDNA was reverse transcribed from 1µg of total RNA extracted from the left ventricular myocardium of male mice (n = 6 for each group) with the primers as shown in table 2. GAPDH and actin genes were used as a reference for normalization of obtained measurements. Briefly, 40 ng of genomic DNA from mouse tail was used to quantify the number of transgenes inserted into the genome. Analysis of gene expression was performed using 2(-delta delta C(T)) method. [54].

Immunoblotting and Histopathology of Myocardium

Picrosirius red staining for assessment of fibrosis was performed by the Research Animal Diagnostic Laboratory (University of Missouri). To determine fibrosis, 5 independent high-power fields of stained images from each animal were analyzed by a blinded observer with Image-Pro Plus software (MediaCybernetics, Silver Spring, MD).

Statistics

All results are expressed as means ± SE. Two-way analysis of variance was used to analyze the calcium transient. Repeated measurement ANOVA was used to analyze the contractile function after TAC. Commercial software package were used for all statistical analysis (Graph Pad, La Jolla, CA) two group comparisons were made with the unpaired student t-test. In all analyses, p<0.05 was taken to be statistically significant.

Author Contributions

Conceived and designed the experiments: EAH WZZ TOC AMF. Performed the experiments: EAH WZZ TOC VM EHG XL JZ JLS XQZ. Analyzed the data: EAH WZZ TOC JYC WK AMF. Contributed reagents/materials/analysis tools: VM JZ. Wrote the paper: WZZ TOC AFM.

References

  1. 1. Headrick JP HB, Ashton KJ (2003) Acute adenosinergic cardioprotection in ischemic-reperfused hearts. Am J Physiol Heart Circ Physiol 285: H1797–H1818.
  2. 2. Sullivan GW RJ, Scheld WM, Macdonald TL, Linden J (2001) Cyclic AMP-dependent inhibition of human neutrophil oxidative activity by substituted 2-propynylcyclohexyl adenosine A(2A) receptor agonists. Br J Pharmacol 132: 1017–1026.
  3. 3. Dobson JG Jr, Fenton RA (1997) Adenosine A2 receptor function in rat ventricular myocytes. Cardiovasc Res 34: 337–347.
  4. 4. Headrick JP WL, Ashton KJ, Holmgren K, Peart J, Matherne GP (2003 ) Ischaemic tolerance in aged mouse myocardium: the role of adenosine and effects of A1 adenosine receptor overexpression. J of physiol 549 (pt 3): 823–833.
  5. 5. Safran N SV, Balas N, Jacobson KA, Nawrath H, Shainberg A (2001) Cardioprotective effects of adenosine A1 and A3 receptor activation during hypoxia in isolated rat cardiac myocytes. Mol Cell Biochem 217: 14.
  6. 6. Todd J, Zhao ZQ, Williams MW, Sato H, Van Wylen DG, et al. (1996) Intravascular adenosine at reperfusion reduces infarct size and neutrophil adherence. Ann Thorac Surg 62: 1364–1372.
  7. 7. Vinten-Johansen J, Thourani VH, Ronson RS, Jordan JE, Zhao ZQ, et al. (1999) Broad-spectrum cardioprotection with adenosine. Ann Thorac Surg 68: 1942–1948.
  8. 8. Dubey RK, Gillespie DG, Mi Z, Jackson EK (1997) Exogenous and endogenous adenosine inhibits fetal calf serum-induced growth of rat cardiac fibroblasts: role of A2B receptors. Circulation 96: 2656–2666.
  9. 9. Wagner DR, McTiernan C, Sanders VJ, Feldman AM (1998) Adenosine inhibits lipopolysaccharide-induced secretion of tumor necrosis factor-alpha in the failing human heart. Circulation 97: 521–524.
  10. 10. Chung ES, Perlini S, Aurigemma GP, Fenton RA, Dobson JG Jr, et al. (1998) Effects of chronic adenosine uptake blockade on adrenergic responsiveness and left ventricular chamber function in pressure overload hypertrophy in the rat. J Hypertens 16: 1813–1822.
  11. 11. Liao Y, Takashima S, Asano Y, Asakura M, Ogai A, et al. (2003) Activation of adenosine A1 receptor attenuates cardiac hypertrophy and prevents heart failure in murine left ventricular pressure-overload model. Circ Res 93: 759–766.
  12. 12. Gan XT, Rajapurohitam V, Haist JV, Chidiac P, Cook MA, et al. (2005) Inhibition of phenylephrine-induced cardiomyocyte hypertrophy by activation of multiple adenosine receptor subtypes. J Pharmacol Exp Ther 312: 27–34.
  13. 13. Xu X, Fassett J, Hu X, Zhu G, Lu Z, et al. (2008) Ecto-5′-nucleotidase deficiency exacerbates pressure-overload-induced left ventricular hypertrophy and dysfunction. Hypertension 51: 1557–1564.
  14. 14. Lu Z, Fassett J, Xu X, Hu X, Zhu G, et al. (2008) Adenosine A3 receptor deficiency exerts unanticipated protective effects on the pressure-overloaded left ventricle. Circulation 118: 1713–1721.
  15. 15. Funakoshi H, Chan TO, Good JC, Libonati JR, Piuhola J, et al. (2006) Regulated overexpression of the A1-adenosine receptor in mice results in adverse but reversible changes in cardiac morphology and function. Circulation 114: 2240–2250.
  16. 16. Hunter JJ, Chien KR (1999) Signaling pathways for cardiac hypertrophy and failure. N Engl J Med 341: 1276–1283.
  17. 17. Jessup M, Brozena S (2003) Heart failure. N Engl J Med 348: 2007–2018.
  18. 18. Harada K, Sugaya T, Murakami K, Yazaki Y, Komuro I (1999) Angiotensin II type 1A receptor knockout mice display less left ventricular remodeling and improved survival after myocardial infarction. Circulation 100: 2093–2099.
  19. 19. Gajewski K, Fossett N, Molkentin JD, Schulz RA (1999) The zinc finger proteins Pannier and GATA4 function as cardiogenic factors in Drosophila. Development 126: 5679–5688.
  20. 20. Amedeo Modesti P, Zecchi-Orlandini S, Vanni S, Polidori G, Bertolozzi I, et al. (2002) Release of preformed Ang II from myocytes mediates angiotensinogen and ET-1 gene overexpression in vivo via AT1 receptor. J Mol Cell Cardiol 34: 1491–1500.
  21. 21. Sanbe A, Gulick J, Hanks MC, Liang Q, Osinska H, et al. (2003) Reengineering inducible cardiac-specific transgenesis with an attenuated myosin heavy chain promoter. Circ Res 92: 609–616.
  22. 22. Hamad EA, Li X, Song J, Zhang XQ, Myers V, et al. (2010) Effects of cardiac-restricted overexpression of the A(2A) adenosine receptor on adriamycin-induced cardiotoxicity. Am J Physiol Heart Circ Physiol 298: H1738–1747.
  23. 23. Chan TO, Funakoshi H, Song J, Zhang XQ, Wang J, et al. (2008) Cardiac-restricted overexpression of the A(2A)-adenosine receptor in FVB mice transiently increases contractile performance and rescues the heart failure phenotype in mice overexpressing the A(1)-adenosine receptor. Clin Transl Sci 1: 126–133.
  24. 24. Funakoshi H, Zacharia LC, Tang Z, Zhang J, Lee LL, et al. (2007) A1 adenosine receptor upregulation accompanies decreasing myocardial adenosine levels in mice with left ventricular dysfunction. Circulation 115: 2307–2315.
  25. 25. Xia Y, Lee K, Li N, Corbett D, Mendoza L, et al. (2009) Characterization of the inflammatory and fibrotic response in a mouse model of cardiac pressure overload. Histochem Cell Biol 131: 471–481.
  26. 26. Nagai T, Anzai T, Kaneko H, Mano Y, Anzai A, et al. (2011) C-reactive protein overexpression exacerbates pressure overload-induced cardiac remodeling through enhanced inflammatory response. Hypertension 57: 208–215.
  27. 27. Higuchi Y, Chan TO, Brown MA, Zhang J, DeGeorge BR Jr, et al. (2006) Cardioprotection afforded by NF-kappaB ablation is associated with activation of Akt in mice overexpressing TNF-alpha. Am J Physiol Heart Circ Physiol 290: H590–598.
  28. 28. Hasko G, Pacher P (2008) A2A receptors in inflammation and injury: lessons learned from transgenic animals. J Leukoc Biol 83: 447–455.
  29. 29. Impellizzeri D, Di Paola R, Esposito E, Mazzon E, Paterniti I, et al. (2011) CGS 21680, an agonist of the adenosine (A2A) receptor, decreases acute lung inflammation. Eur J Pharmacol 668: 305–316.
  30. 30. Pinhal-Enfield G, Ramanathan M, Hasko G, Vogel SN, Salzman AL, et al. (2003) An angiogenic switch in macrophages involving synergy between Toll-like receptors 2, 4, 7, and 9 and adenosine A(2A) receptors. Am J Pathol 163: 711–721.
  31. 31. Grossman W, Jones D, McLaurin LP (1975) Wall stress and patterns of hypertrophy in the human left ventricle. J Clin Invest 56: 56–64.
  32. 32. Grossman W, McLaurin LP, Stefadouros MA (1974) Left ventricular stiffness associated with chronic pressure and volume overloads in man. Circ Res 35: 793–800.
  33. 33. Opie LH, Commerford PJ, Gersh BJ, Pfeffer MA (2006) Controversies in ventricular remodelling. Lancet 367: 356–367.
  34. 34. Harris DE, Work SS, Wright RK, Alpert NR, Warshaw DM (1994) Smooth, cardiac and skeletal muscle myosin force and motion generation assessed by cross-bridge mechanical interactions in vitro. J Muscle Res Cell Motil 15: 11–19.
  35. 35. Holubarsch C, Goulette RP, Litten RZ, Martin BJ, Mulieri LA, et al. (1985) The economy of isometric force development, myosin isoenzyme pattern and myofibrillar ATPase activity in normal and hypothyroid rat myocardium. Circ Res 56: 78–86.
  36. 36. Holubarsch C, Litten RZ, Mulieri LA, Alpert NR (1985) Energetic changes of myocardium as an adaptation to chronic hemodynamic overload and thyroid gland activity. Basic Res Cardiol 80: 582–593.
  37. 37. Krenz M, Robbins J (2004) Impact of beta-myosin heavy chain expression on cardiac function during stress. J Am Coll Cardiol 44: 2390–2397.
  38. 38. Dorn GW 2nd, Robbins J, Ball N, Walsh RA (1994) Myosin heavy chain regulation and myocyte contractile depression after LV hypertrophy in aortic-banded mice. Am J Physiol 267: H400–405.
  39. 39. Yan X, Schuldt AJ, Price RL, Amende I, Liu FF, et al. (2008) Pressure overload-induced hypertrophy in transgenic mice selectively overexpressing AT2 receptors in ventricular myocytes. Am J Physiol Heart Circ Physiol 294: H1274–1281.
  40. 40. Charron F, Tsimiklis G, Arcand M, Robitaille L, Liang Q, et al. (2001) Tissue-specific GATA factors are transcriptional effectors of the small GTPase RhoA. Genes Dev 15: 2702–2719.
  41. 41. Liang Q, De Windt LJ, Witt SA, Kimball TR, Markham BE, et al. (2001) The transcription factors GATA4 and GATA6 regulate cardiomyocyte hypertrophy in vitro and in vivo. J Biol Chem 276: 30245–30253.
  42. 42. Diedrichs H, Chi M, Boelck B, Mehlhorn U, Schwinger RH (2004) Increased regulatory activity of the calcineurin/NFAT pathway in human heart failure. Eur J Heart Fail 6: 3–9.
  43. 43. Hall JL, Grindle S, Han X, Fermin D, Park S, et al. (2004) Genomic profiling of the human heart before and after mechanical support with a ventricular assist device reveals alterations in vascular signaling networks. Physiol Genomics 17: 283–291.
  44. 44. Yang D, Ma S, Tan Y, Li D, Tang B, et al. (2010) Adrenergic receptor blockade-induced regression of pressure-overload cardiac hypertrophy is associated with inhibition of the calcineurin/NFAT3/GATA4 pathway. Mol Med Report 3: 497–501.
  45. 45. Morimoto T, Hasegawa K, Wada H, Kakita T, Kaburagi S, et al. (2001) Calcineurin-GATA4 pathway is involved in beta-adrenergic agonist-responsive endothelin-1 transcription in cardiac myocytes. J Biol Chem 276: 34983–34989.
  46. 46. Morisco C, Seta K, Hardt SE, Lee Y, Vatner SF, et al. (2001) Glycogen synthase kinase 3beta regulates GATA4 in cardiac myocytes. J Biol Chem 276: 28586–28597.
  47. 47. Saadane N, Alpert L, Chalifour LE (1999) Expression of immediate early genes, GATA-4, and Nkx-2.5 in adrenergic-induced cardiac hypertrophy and during regression in adult mice. Br J Pharmacol 127: 1165–1176.
  48. 48. Csoka B, Nemeth ZH, Virag L, Gergely P, Leibovich SJ, et al. (2007) A2A adenosine receptors and C/EBPbeta are crucially required for IL-10 production by macrophages exposed to Escherichia coli. Blood 110: 2685–2695.
  49. 49. De Ponti C, Carini R, Alchera E, Nitti MP, Locati M, et al. (2007) Adenosine A2a receptor-mediated, normoxic induction of HIF-1 through PKC and PI-3K-dependent pathways in macrophages. J Leukoc Biol 82: 392–402.
  50. 50. Fiser SM, Tribble CG, Kaza AK, Long SM, Kern JA, et al. (2002) Adenosine A2A receptor activation decreases reperfusion injury associated with high-flow reperfusion. J Thorac Cardiovasc Surg 124: 973–978.
  51. 51. McPherson JA, Barringhaus KG, Bishop GG, Sanders JM, Rieger JM, et al. (2001) Adenosine A(2A) receptor stimulation reduces inflammation and neointimal growth in a murine carotid ligation model. Arterioscler Thromb Vasc Biol 21: 791–796.
  52. 52. Okusa MD, Linden J, Macdonald T, Huang L (1999) Selective A2A adenosine receptor activation reduces ischemia-reperfusion injury in rat kidney. Am J Physiol 277: F404–412.
  53. 53. Most P SH, Gao E, Funakoshi H, Volkers M, Heierhorst J, et al. (2006) Cardiac S100A1 protein levels determine contractile performance and propensity toward heart failure after myocardial infarction. Circulation 114: 1258–1268.
  54. 54. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402–408.